You are on page 1of 17

Article

Critical issues in tissue engineering:


biomaterials, cell sources, angiogenesis,
and drug delivery systems

Journal of Biomaterials Applications


0(0) 117
! The Author(s) 2011
Reprints and permissions:
sagepub.co.uk/journalsPermissions.nav
DOI: 10.1177/0885328211408946
jba.sagepub.com

Hojjat Naderi1, Maryam M Matin1,2 and Ahmad Reza Bahrami1,2

Abstract
Tissue engineering is a newly emerging biomedical technology, which aids and increases the repair and regeneration of
deficient and injured tissues. It employs the principles from the fields of materials science, cell biology, transplantation,
and engineering in an effort to treat or replace damaged tissues. Tissue engineering and development of complex tissues
or organs, such as heart, muscle, kidney, liver, and lung, are still a distant milestone in twenty-first century. Generally,
there are four main challenges in tissue engineering which need optimization. These include biomaterials, cell sources,
vascularization of engineered tissues, and design of drug delivery systems. Biomaterials and cell sources should be specific
for the engineering of each tissue or organ. On the other hand, angiogenesis is required not only for the treatment of a
variety of ischemic conditions, but it is also a critical component of virtually all tissue-engineering strategies. Therefore,
controlling the dose, location, and duration of releasing angiogenic factors via polymeric delivery systems, in order to
ultimately better mimic the stem cell niche through scaffolds, will dictate the utility of a variety of biomaterials in tissue
regeneration. This review focuses on the use of polymeric vehicles that are made of synthetic and/or natural biomaterials
as scaffolds for three-dimensional cell cultures and for locally delivering the inductive growth factors in various formats
to provide a method of controlled, localized delivery for the desired time frame and for vascularized tissue-engineering
therapies.
Keywords
tissue engineering, scaffold, biomaterials, cell sources, angiogenesis, drug delivery systems

Introduction
There are serious limitations in using either autologous
or allogeneic grafts in traditional transplantation surgeries. These include lack of appropriate donor tissues
in autologous transplantation, and risk of disease transmission and extended immune-rejection in allogeneic
transplantation. However, tissue engineering oers to
circumvent these problems by replacing and restoring
various tissues and organs through the delivery of stem
cells (SCs) and bioactive molecules onto specic biomaterials in a three dimensional (3D) structure.1
The cell microenvironment is known to play a
remarkable role in determining progenitor cell fate
and function. The exact coordination of spatial and
temporal cues from the microenvironment is highly
essential for SCs to constitute complex functional tissues. Advanced and high throughput assays, such as
extracellular matrix (ECM) microarrays and other
technologies have unveiled specic cell interactions

with ECM components and polymers that can inuence


SC signaling and response.26 As long as biomaterial
research moves forward, new materials and innovations
in their manipulation and usage will continue to
emerge. By utilizing high throughput arrays to recognize the function of biomaterials, the evaluation of
these molecules for ultimate usage can take place in
short periods of time.5,7,8 So, major advances in both
the understanding of the local cues required for lineage
commitment and the discovery of new biomaterials necessary to support SCs and drug delivery can be advantageous. The promise of cell therapy lies in the repair of
1

Department of Biology, Ferdowsi University of Mashhad, Mashhad, Iran


Cell and Molecular Biology Research Group, Institute of Biotechnology,
Ferdowsi University of Mashhad, Mashhad, Iran
2

Corresponding author:
Maryam M. Matin, Department of Biology, Faculty of Science, Ferdowsi
University of Mashhad, Mashhad, Iran
Email: matin@um.ac.ir

Downloaded from jba.sagepub.com at University of Liverpool on March 27, 2016

Journal of Biomaterials Applications 0(0)

damaged tissues and organs in vivo and also the


production of appropriate tissues in vitro for successful
transplantation. In the recent years, in order to mimic
the SC niche, a variety of biomaterials have been combined with SC cultures, to prepare a suitable microenvironment for their growth and dierentiation.2,911
The reconstruction and direct replacement of diseased cells and tissues are becoming a clinical possibility mainly because of correlated advances in the
modication of biomaterials and comprehension of
SCs behavior.1214 Creation of cell-compatible biomaterial microarrays has allowed rapid, microscale testing
of biomaterial interactions with cells.2,6 Several review
articles have discussed how specic types of biomaterials are used as substances to mimic the physicochemical microenvironments of cells and tissues.1416
A number of studies have specically examined materials for the development of bone,17,18 cartilage,19
skin,20 and blood vessels.21 Others have reported studies on various pre-dierentiated SC populations and
their combination together using biomaterials to form
hybrid constructs that closely mimic native tissues.2224
However, the eld of tissue engineering is restricted
by the necessity for angiogenesis in large tissues and
organs for nutrient, waste, and oxygen transport.
Strategies to induce new blood vessel networks will
be essential in almost all engineered tissues.25,26
There have been many reports on the design and
improvement of scaold materials to help local angiogenesis directly in vivo and promote gradual penetration of host vessels into the scaolds.27,28 Even for
ex vivo pre-vascularized scaolds, successful joining

of the graft with the host tissues largely relies on


vessel and tissue integration. One major theme guiding
this approach is delivery of angiogenic factors from
implanted scaolds, which will be discussed in this
review. While systemic injection of angiogenic molecules such as vascular endothelial growth factor
(VEGF) is counterpart with negative side eects in
non-target tissues (hyperpermeable vessels, hypotension, motivation of tumor growth, and unhindered
neovascularization), prolonged delivery of angiogenic
factors from scaold materials can be localized to a
microenvironment to minimize the negative side eects
in non-target regions. In addition, natural processes of
secretion and sequestration of angiogenic factors in
ECM beds can be imitated in this system by adjusting
their release kinetics from the scaolds.2931
In spite of the good progress in bioengineering of
tissues composed of thin layers of cells, such as skin,
a main dispute for future tissue engineering is the creation of larger organs with more complex structures,
such as kidney, heart, and liver. Tissues with a huge
mass of cells require a vascular network of capillaries,
arteries, and veins for the transport of nutrients and
oxygen to each cell. Development of eective methods
for angiogenesis of these tissues is critical for obtaining
a successful outcome.32 In this review, we will strive to
describe major advances in the eld of tissue engineering, including biomaterials, cell sources, engineering
of thick tissues or organs, drug delivery systems
(DDSs, Figure 1), and novel ndings to better mimic
the native tissues for preclinical and clinical tissue
engineering.

Critical challenges in tissue engineering

1-Biomaterials

Origin

Natural

Synthetic

Stem cells

Types

Composite

3-Angiogenesis of
engineered tissues

2-Cell sources

Injectable

In situ
forming
hydrogels,
Glues, Selfassembling
peptides

Noninjectable

ESCs,
MSCs,
EPCs,
HSCs

Differentiated
cells

With
signaling
molecules

Use of VEGF,
bFGF, PDGF.
and TGF-
alone

With
endothelial
cells (ECs)

Use of multiple
growth factors

4-Drug delivery
systems
Biomaterialbased

Microfluidic
devices

Microspheres,
Nanospheres,
Incorporation,
Microparticle beads,
Patterning devices

Porous scaffolds,
Decellularized
ECM, Meshs, ECM
secreting cell
sheets, Gels,
Sponges, Sutures,
Microspheres,
Nanofibres

Figure 1. Overview of critical issues in tissue engineering.


Notes: ESC, embryonic stem cell; MSC, mesenchymal stem cell; EPC, endothelial progenitor cell; HSC, hematopoietic stem cell; VEGF,
vascular endothelial growth factor; bFGF, basic fibroblast growth factor; PDGF, platelet-derived growth factor; and TGF-b, transforming growth factor-b.

Downloaded from jba.sagepub.com at University of Liverpool on March 27, 2016

Naderi et al.

Although classical two dimensional (2D) cell cultures are widely used and have provided many advances
in cell biology, but due to the fact that cells (including
SCs) reside, proliferate, migrate, and dierentiate inside
the body within complicated 3D microenvironments,
most of the current research in biomaterial-directed
SCs manipulation is focused on such 3D environments.11 This review will primarily focus on the concept
of 3D culture, biomaterials and their modications, and
later on we will discuss other important issues in the
engineering of thick tissues.

Biomaterials
The term biomaterials has many denitions; one traditional meaning indicates that a biomaterial is a nonliving substance used in a medical device, like a joint
prosthesis. However, the technology of biomaterials
has developed gradually, and the expanded denition
includes substances that are designed to control the biological environment of cells and tissues. More than being
simply compatible with the host and serving a structural
role, biomaterials can now direct cells through microenvironmental cues.33
Biomaterial-based 3D systems have been the most
inuential tools in rendering a scaold to cells, both in
culture or inside the body. These 3D structures present
an ideal substrate for cellcell and cellmaterial
communications, and their properties can be modied
to induce dierentiation of cells into specic lineages.34
Scaolds used for tissue engineering perform many
functions and their role during tissue formation is dependent on the specic characteristics of the selected
biomaterials.35 It has been proven that 3D scaolds
enhance osteogenic,36 hematopoietic,37 neural,38,39 and
chondrogenic40 dierentiation. Thus, in addition to
acting as delivery vehicles for biomolecules during
tissue development,9 biomaterials promote cell attachment, proliferation, organization, and dierentiation.41
Properties of biocompatible scaolds, synthetic or
natural, can be considered from dierent aspects including optimal nutrient and waste transport, delivery of bioactive molecules, material degradation rate, cellrecognizable surface chemistries, mechanical unity, and
the ability to promote signal transduction pathways. The
considerable success of tissue organization and development highly depends on these properties, because they
can eventually dictate cell adherence, nutrient/waste
transport, cell dierentiation, cell viability, and matrix
synthesis and organization. Most of the materials in
scaolds can be chemically or physically modied to
control all these important parameters, and a variety of
synthetic and natural materials have been used for investigating SCs behavior by specically manipulating these

properties. Several articles have reviewed the application


of scaolds in tissue engineering in general.42,43 In an
optimal form, a biomaterial must degrade without
toxicity and with a controlled degradation rate.
Contrary to a constantly implanted structural prosthesis,
scaold biomaterials should remain long enough to conduct joining of recruited or applied cells, but not persist
so long as to obstruct the nal cellcell physiological
coupling necessary for tissue engineering.
An important property of biomaterials is the degradation rate. A rapid degradation can jeopardize the
mechanical unity of biomaterials. Therefore, it is desirable to control degradation and stiness independently.
Diverse approaches can regulate biomaterials degradation. The molecular weight and copolymerization ratio
can be easily controlled to optimize the degradation
rate.4446 Kong et al. showed that modifying alginates
with various cross-linking strategies could keep stiness
but increase degradation, improving bone formation by
bone marrow-derived mesenchymal stem cells.47 Thus,
degradation of polymers can potentially be adjusted for
the regenerative strategy as long as transiently supporting mechanical integrity exists.
Another important feature of scaolds is porosity.
The pore size (both length and cross-sectional area),
pore numbers, and pore connectivity of scaolds are
key factors in determining their function. Size of
pores on a length scale of micrometers to millimeters
strongly aects tracking of cells; extremely large
pores could spoil vascularization, since endothelial
cells (ECs) are not capable of bridging pores larger
than a cell diameter.48 In contrast, pores smaller than
100 nm will inuence diusion of nutrients, waste, and
oxygen. Poor diusion of factors and nutrients may
result in the failure of implant and reduced survival
of implanted cells. Porosity needs to be balanced with
the integrity of the materials, their mechanical properties and cellular eects.4951 Some hydrogels, like those
formed by self-assembling peptides, have very small
pore sizes that encourage endothelial adhesion and capillary formation, but still permit rapid cell migration
because of the hydrogels exibility.52
There are natural, synthetic, and composite materials
that can be injectable or non-injectable (Figure 1).
Injectable polymers are important biomaterials, because
of their clinical applicability without surgery, as DDSs
for tissue engineering.28,5356 Several synthetic and
natural biodegradable polymers, including polyesters,
poly(amino acids), polysaccharides, and proteins have
been studied well.53,57,58 In addition, chemical and
biological modications of materials can result in better
mimicking the SCs niche and create specic microenvironments to control cell responses.11 Dierent types of
biomaterials and their modications are discussed, in
more details, in the following sections.

Downloaded from jba.sagepub.com at University of Liverpool on March 27, 2016

Journal of Biomaterials Applications 0(0)

Natural biomaterials
Natural biomaterials used for scaolds include components found in the ECM such as collagen, brinogen,
hyaluronic acid, glycosaminoglycans (GAGs), and
hydroxyapatite (HA), and therefore have the benet
of being bioactive, biocompatible, and having mechanical properties that are common with native tissues.57
Furthermore, other natural materials including those
derived from plants, insects, or animals (e.g., cellulose,
chitosan, silk broin, etc.) can provide favorable microenvironments for the culture of SCs.59,60 Drawbacks of
using natural materials rather than synthetic materials
include restricted control over their physico-chemical
properties, inability to moderate their degradation
rates, challenges in sterilization and purication techniques, and also pathogen/viral issues when extracting
from dierent sources.61
There are many natural biomaterials used as 3D
scaolds for tissue engineering. Several natural materials, e.g., chitosan, Matrigel, hyaluronic acid, and
brin, which have become commercially available, are
well characterized, and have reproducible, controlled
properties. Chitosan, as an ideal scaold, has been
widely used in the tissue engineering of skin, bone, cartilage, liver, nerve, blood vessels, and heart in the past
25 years.55 Chitosan has been approved by the US
Food and Drug Administration and is used in drug
delivery and tissue engineering. Derivatives of chitosan
including porous structures, chitosan-based nanobrous structures, and injectable chitosan hydrogels
have dierent applications in tissue engineering.
Chitosan hydrogel responds to a variety of external
stimuli such as pH, light, and temperature. The temperature-responsive chitosan in combination with glycerol phosphate (GP) as injectable hydrogel is highly
attractive and has great applications, because bioactive
factors (such as growth factors, genes, and supportive
cells relevant to the repair and regeneration of the
tissues) can be easily incorporated into the polymer
solution.56 Then, once exposed to body temperature
(37 C), the polymer solution can polymerize rapidly
in situ within a short time, trapping these factors
within the injected area. This ability for in situ
polymerization makes chitosan-GP a clinically useful
scaold.55,56,62
Commercially available MatrigelTM is a complex
protein mixture including laminin, collagen IV, and
heparan sulfate proteoglycans.63 Laschke et al. demonstrated that the incorporation of Matrigel into
poly(lactide-co-glycolide) (PLGA) scaolds can accelerate adequate vascularization of tissue engineering
constructs.64 Other researchers showed that when
human endothelial progenitor cells (hEPCs) were incorporated into Matrigel, and implanted subcutaneously

into immunodecient mice, vascular networks were created in vivo.65


Hyaluronic acid is another attractive natural biomaterial, which is involved in cell signaling and behavior.
Although hyaluronic acid is present in tissues as a gellike substance, it can be chemically modied for eective
processing into bers, membranes, or microspheres. An
altered type of hyaluronic acid is commercially available
as Hya .66 Hya -based scaolds are biodegradable
and combine the advantages of being a natural material
with allowing the cells to replace the scaold with their
own ECM. Recently, Gerecht et al.67 reported the use of
hyaluronic acid hydrogels for maintaining the pluripotency and undierentiated state of human embryonic
stem cells (hESCs) and showed that addition of soluble
growth factors to these hydrogels could successfully trigger lineage specic dierentiation of ES cells.
Fibrin is another class of natural materials that can
be applied to make 3D scaold materials.68 This scaffold, in conjunction with various growth factors,
showed remarkable increase in neuron production and
neuronal viability39 and also in clinical cartilage engineering.69 Fibrin glue is a synthetic substance used to
create a brin clot. It is made of brinogen and thrombin. Thrombin acts as an enzyme that converts brinogen into brin in 1060 s, so that brin can be used as
injectable in situ forming gel. Fibrin scaolds have been
used for injection in a preclinical ischemic heart study.70

Synthetic biomaterials
Poly(glycolic acid) (PGA), poly(lactic acid) (PLA), and
the copolymer PLGA have been extensively used as synthetic 3D scaold biomaterials for assessing cell behavior.7173 Necessary criteria such as biocompatibility,
processability, and controlled degradation are fullled
with these polyesters.74 These biomaterials degrade
hydrolytically via mass erosion and the glycolic/lactic
acid byproducts are physiologically removed through
metabolic pathways. The molecular weight, copolymerization ratio, and polydispersity of the polymers can be
easily tunable to control the degradation rate. These
properties have made synthetic materials highly attractive for tissue engineering. In addition, standard processing methods (e.g., salt leaching, sintering, porogen
melting, and nanober electrospinning) have been well
established to prepare a wide variety of 3D scaolds
using synthetic biomaterials.75,76
Synthetic materials provide the versatility of creating
3D microenvironments with adjustable features
including mechanical properties, degradation rates, and
porosity. However, in spite of these benets, they have
poor inherent bioactivity (e.g., polyethylene glycol,
PEG), acidic byproducts (e.g., PGA, PLA, or PLGA),
etc. Therefore, it is critical to modify synthetic materials

Downloaded from jba.sagepub.com at University of Liverpool on March 27, 2016

Naderi et al.

with biological or chemical compounds to obtain suitable cellular responses. The physical properties of these
polymers can also be easily controlled by changing the
ratio of lactide:glycolide, molecular weight, and their
crystallinity.74,77
Use of composite scaolds is another approach to
better mimic physiological niche and improve the process of tissue engineering.46,78 Natural or synthetic
hydrogels closely resemble the consistency of soft,
native tissues, making them attractive scaold materials
for soft tissue engineering. On the other hand, sometimes composite materials with higher mechanical
strength are required to closely mimic the tissue
mechanical properties and optimize degradation rate.
For example, hydrogel-like materials can be modied
to have increased elasticity, making them more suitable
for applications in connective tissue engineering.
Collagen gels can also be adjusted to have a higher
elasticity by adding HA. Calcium HA is the main component of teeth and bones in vertebrates, thereby imitating the composition of bone, which is mainly
composed of collagen bers and phosphate minerals.79
Zawaneh et al. have reported the design of an injectable synthetic and biodegradable polymeric biomaterial
comprising polyethylene glycol and a polycarbonate
of dihydroxyacetone (MPEGPDHA) that is easily
extruded through narrow-gage needles, biodegrades
into inert products and is well tolerated by soft tissues.
This type of polymer holds signicant promise for
clinical applications in patients going through surgical
procedures ranging from cosmetic surgery to cancer
resection and tissue engineering.53

Modification of biomaterials
Physical, chemical, and biological modications of biomaterials can directly inuence SCs behavior by altering
scaold properties, surface interactions, scaold degradation rate, microenvironmental architecture, and
manipulating the signal transduction pathways in SCs.
Biomaterials can be designed to precisely control their
degradation kinetics, present specic ligand-based
signals, and/or control the release of biomolecules in
response to the microenvironment.77 These can inuence
cellmatrix interactions and may lead to altered gene
expression and lineage specicity. Many studies have
demonstrated how modied biomaterials and scaold
surface properties introduce specic biological responses
in SCs.34,77,8083 Therefore, the goal of biomaterial-directed SC culture is to mimic physical and biochemical properties of the physiological SC niche.77,84
When anchorage-dependent cells are cultured on
various biomaterials, ECM proteins including collagen,
bronectin, hyaluronic acid, GAGs, brin, and gelatin
are generally used to cover surfaces of various

biomaterials to enhance their interactions with cells.


Cellular interactions with ECM proteins are very
complicated because these proteins contain multiple
cell- and growth factor-binding domains. To avoid
these problems, short peptides only several amino acids
in length, have been derived from ECM proteins as the
most primitive subunits needed for normal cell attachment and proliferation. Employment of synthetic peptides in cell cultures and engineered tissues can
overcome the need for bulk production and purication
of ECM proteins from tissue extracts. Holtorf et al. evaluated titanium ber mesh scaolds coated with Arg-GlyAsp (RGD) sequence, a cell adhesive, integrin-binding
peptide found in bronectin and laminin, and showed
that mesenchymal stem cells (MSCs) could attach more
strongly to these RGD-coated scaolds.85 Cellmatrix
interactions were enhanced in RGD functionalized
hydrogels, resulting in increased MSCs viability.
Nuttelman et al.86 showed that the viability of the encapsulated hMSCs augmented from 15% to 75% when
RGD was added into PEG-diacrylate hydrogels. Also,
hESCs cultured on this completely synthetic ECM substitute were shown to be morphologically similar to
hESCs cultured on an embryonic broblast feeder layer
and generated markers typical of undierentiated ES
cells.87 Many studies have reported that RGD sequence
helps in the attachment of various cell types including
broblasts,88 smooth muscle cells (SMCs),89 preosteoblasts,90 preadipocytes,91 and MSCs.92 Specially, ECs
have been favorably cultured on RGD-containing polymers including hyaluronic acid hydrogels,93 derivatives
of isopropylacrylamides,94 and PEG hydrogels.95
Tyr-Iso-Gly-Ser-Arg (YIGSR), a laminin-derived
short peptide sequence, combined with polyurethanes,
could selectively enhance ECs adhesion and proliferation but decreased platelet adhesion.96 Glass97 and
PEG hydrogels98 coated with YIGSR also increased
EC attachment and migration. Arg-Glu-Asp-Val
(REDV) sequence originated from bronectin, binds
to integrins found on ECs, supporting specic adhesion
of these cells.99 Recombinant ECM proteins with
REDV sequence were constructed and applied as vascular graft biomaterials to encourage trapping of
ECs.100 These studies have shown that short peptides
can replace massive ECM proteins as coating materials
and enhance cellular adhesion and functions on biomaterials. Furthermore, these short peptides with adhesive
properties could be micropatterned into specic regions
to control spatial arrangement of ECs, and other tissuespecic cell types.95,101,102
SCs dierentiation can be directly mediated by exposure to proper biological or chemical signals in their
microenvironment. It is well established that specic
growth factors, hormones, and cytokines can increase
proliferation and lineage-specic dierentiation of SCs.

Downloaded from jba.sagepub.com at University of Liverpool on March 27, 2016

Journal of Biomaterials Applications 0(0)

For example, broblast growth factor-2 (FGF-2) has


been shown to increase self-renewal of MSCs and maintain their multi-lineage dierentiation capability.103
Moreover, bone morphogenetic proteins (BMPs) have
been shown to play a signicant role in the regeneration
of specic cell types including skeletal tissues, especially
bone.104
Growth factors, hormones, and chemicals have classically been directly added into the culture medium. On
the other hand, these biomolecules can be directly incorporated within the scaold structure or encapsulated
into the scaold biomaterials in a variety of ways
during the scaold manufacturing process.105,106 A
highly used method for the delivery of growth/chemotactic factors in tissue engineering is simple physical
adsorption of biomolecules on biomaterials or scaolds
surface.35 The chemotactic factors such as stromal cellderived factor 1 (SDF1) incorporated in PLGA scaolds
can attract MSCs to the site of implant. Enhanced
homing of autologous MSCs improved the tissue
responses to biomaterial implants through modifying/
bypassing inammatory responses and jumpstarting
SCs participation in healing at the implant interface.71
Covalent immobilization of soluble signaling proteins on biomaterials enables prolonged signaling by
intervening with their endocytosis. By immobilization
technique, bioactivity of the proteins can be retained on
biomaterials. An important angiogenic factor, VEGF,
was covalently incorporated into collagen gels. When
this scaold was implanted on chicken chorioallantoic
membrane, the results indicated improved capillary formation and tissue ingrowth.107,108 It was also shown
that genetically modied VEGF, in which N-terminal
cysteine was conjugated to brin matrix via thiol-directed bifunctional cross-linking reagent, could retain bioactivity, and improved angiogenic performance.109
Another powerful angiogenic growth factor is basic
broblast growth factor (bFGF) that has been immobilized into PEG hydrogels with concentration gradient. The resulting materials could direct cell alignment
and migration of SMCs.110 Cellcell interaction proteins including ephrin-B2111 and ephrin-A1112, which
were incorporated into brin matrices and PEG hydrogels, respectively, showed signicant roles in the induction of angiogenesis. Therefore, the use of special
signaling proteins presents opportunities in the design
of angiogenic biomaterials.

Cell sources
Various sources of SCs and dierentiated cells are used
for tissue engineering as for dierent target tissues
(Figure 1). SCs represent an important building block
for regenerative medicine and tissue engineering. These
cells are broadly classied into embryonic stem cells

(ESCs) and adult SCs. ESCs have a higher regenerative


capacity than adult SCs and can be manipulated to differentiate into other cell types.113,114 Use of hESCs is
restricted by ethical problems and the potential to form
teratomas115,116 and the request for autologous grafts
has made adult progenitor cells more appropriate for
tissue engineering.117 It has also been shown that
hESCs can acquire chromosomal abnormalities118,119
and therefore are more potent to tumorogenesis. Adult
SCs are multipotent cells with high plasticity. They have
been isolated from many tissues including bone marrow,
blood, brain, liver, muscle, and skin.120122 Although
adult SCs have a lower plasticity compared with ESCs,
they have been demonstrated to dierentiate into a variety of cell types and have been used for treatment of
various diseases including ischemia, neural degeneration, and diabetes in animal models.123127
Here, we do not aim to present an inclusive review to
explain the characterization of SCs since several outstanding previous reviews are available.117,128132 We
further intend to emphasize how SC technology can
be made applicable and benecial in angiogenesis for
engineering thick tissues and whole organs.
One emerging issue in the area of SC therapy is
homing and engraftment of injected or resident cells
to the site of damaged tissue.133 We investigated the
expression of chemokine receptors (such as CXCR4),
which are involved in homing, on the surface of human
MSCs.134 For the construction of vascularized tissue, it
might be helpful to implant a chemokine (SDF1)-containing scaold in the site of injury in order to recruit
SCs to the site.71,135 This method may prevent formation of a necrotic core, which results from cell death in
the center of scaolds due to lack of nutrients and
oxygen. While the chemokine is released from the scaffold, cells gradually penetrate into the scaold and full
tissue, along with angiogenesis, is formed.

Application of SC technology in engineering


of thick tissues
The ECs, which cover the inside of arteries, veins, and
capillaries, are one of the major players of angiogenesis
process in physiological and pathological conditions.
ECs are involved in thrombo-withstanding eects, regulation of leukocyte interactions, adjustment of blood
ow and vessel tone, and selective permeability to various materials. Vasculogenesis is a process that results
from the dierentiation of endothelial progenitor
cells (EPCs) to form new blood vessels. On the other
hand, angiogenesis applies to the development of
new capillary blood vessels by a process of budding
from pre-existing vessels.136,137 Therefore, whereas
vasculogenesis is restricted to embryogenesis, angiogenesis may develop from ECs and EPCs, which take part

Downloaded from jba.sagepub.com at University of Liverpool on March 27, 2016

Naderi et al.

in the formation of new vessels in normal and pathological


situations
after
birth
as
well
as
embryogenesis.137,138
Since the discovery of EPCs, there has been an interest
in their use in tissue engineering.139 The relative ease of
extracting these cells and their capability to be expanded
in culture for up to 1000 doublings, while holding their
capacity to dierentiate, has resulted in their extended
use in this eld.140 EPCs can be isolated from bone
marrow or peripheral blood. EPCs have the capacity to
dierentiate into ECs in vitro and also integrate into positions of neovascularization in vivo. The dierentiation of
EPCs to ECs was revealed for the rst time in vasculogenesis event.139 Although EPCs exhibit less plasticity
and less capacity for self-renewal than their parent SCs,
they still have the capacity to dierentiate into several cell
types. EPCs dierentiation, in vitro, depends on culture
conditions. VEGF and bronectin can induce the dierentiation of EPCs into ECs.141
Outgrowth endothelial cells (OECs) are EPCs variants that can produce high cell numbers typically
required in vascular tissue engineering applications.
OECs collected from human peripheral blood can be
proliferated more than 20 passages, proposing a very
good supply of autologous ECs.142,143 Blood- or bone
marrow-derived EPCs (OEC type) have been examined
for the formation of blood vessels.144,145 The mechanisms by which these dierent EPC variants give rise
to new vessel formation may dier. OECs are classied
into early and late outgrowth EPCs isolated from the
mononuclear fraction.146 These show dierent proliferation capacities, dierential secretion of angiogenic
cytokines, and have dierent morphologies.146 Hence,
cells with dierent functions are present within the
mononuclear fraction. Early outgrowth EPCs can
enhance neovascularization, mainly by the secretion of
angiogenic cytokines (interleukin-8 and VEGF), while
on the contrary, late outgrowth EPCs, which have a
high proliferative capacity, serve as a source of ECs.
Therefore, early outgrowth EPCs may function as
sources of angiogenic cytokines with minimal incorporation into the vasculature;146148 this paracrine eect
has also been described for MSCs.149,150 While both
early and late outgrowth EPCs are equally ecient in
promoting neovascularization,146 the use of two cell
types is better than a single type.151 Interestingly, early
outgrowth ones were shown to be cells with spindle
shape morphology at rst week of culture, while late
outgrowth ones were shown to be cells with cobblestone
morphology after 23 weeks of culture.143,146,151 In fact,
while many independent investigators have shown
bone marrow cells to incorporate into the vasculature,
there are a few reports showing that bone marrow
cells do not incorporate into growing vessels and
instead form periadventitial accumulations, where they

express angiogenic/arteriogenic cytokines such as


VEGF, monocyte chemoattractant protein-1, and
FGF.145,152154 This may represent various cell populations within the mononuclear fraction, which have different cellular and molecular mechanisms in aecting
neovacularization, and can be used in tissue engineering
alone or in combination.
Some experiments have suggested that blood- and
bone marrow-derived primary EPCs possess extended
plasticity. Melero-Martin et al.65 showed that hEPCs
isolated from human umbilical cord blood or from
adult peripheral blood, which were combined with
Matrigel and implanted subcutaneously into immunodecient mice, showed vasculogenic activity and created vascular networks in vivo. EPCs were also able
to dierentiate into cardiomyocytes when co-cultured
with newborn rat cardiomyocytes155 and could exhibit
a mesenchymal phenotype in response to transforming
growth factor b-1 (TGFb-1).156 In addition, expression
of TGFb-1 in SMCs regulates EPCs migration and
dierentiation.157 Thus, transdierentiation of EPCs
might be possible via signaling molecules. As a result,
one can build biomaterial scaolds engineered with
inductive cues to induce cardiomyocyte formation
from EPCs, increasing low-eciency transdierentiation event to a clinically relevant level.158
Many studies have demonstrated that co-culture of
vascular cell types (ECs, EPCs, or OECs) with secondary supporting cells is very eective in the production
of vascularized tissue constructs.159164 Several reports
have suggested the use of MSCs for recruitment of
EPCs/ECs and promotion of angiogenesis due to
their paracrine eects.149,150 However, MSCs are also
able to dierentiate into multiple lineages including cardiomyocytes and vascular ECs.149
Clinical application of tissue engineering is still limited, due to demands for highly specialized cell culture,
isolation, and enrichment techniques required for this
purpose. More understanding of the proliferation and
dierentiation processes that occur in transplanted SCs
populations within the scaolds will increase our success in tissue engineering. An inceptive area of bioengineering investigations is the development of natural or/
and synthetic biopolymer matrices as specic environments for EPCs recruitment, growth, and dierentiation by providing sites of attachment together with
signals that control EPCs migration, survival, and
propagation with synchronized dierentiation.156 The
combination of phenotypic shifts and further comprehension of progenitor cell behaviors will provide inuential tools in advancing thick tissue engineering. It is
also important to know that various sources of SCs
may have dierent responses in the same scaffold117,120,165 and sometimes various scaolds have
diverse eects on the same type of SCs.10,11,166

Downloaded from jba.sagepub.com at University of Liverpool on March 27, 2016

Journal of Biomaterials Applications 0(0)

Generally, there are three approaches commonly


used for vascularization of tissue constructs, as illustrated in Figure 2.162,167,168 The second and third
approaches are described below.

Induction of angiogenesis by the use of cells


and/or angiogenic factors
In spite of considerable attempts in making functional
tissues and organs, most applications of tissue engineering have been restricted to avascular or thin tissues such
as cartilage, skin, or bladder.169,170 In these tissues,
nutrients and oxygen can diuse into the implants
and retain cellular viability. However, as the tissue
becomes thicker, cells existing more than a few hundred
microns away from nearest capillaries would undergo
hypoxia and apoptosis.9 So, the main obstacle in the
creation of more complex tissues is the formation of
vascular networks able to deliver nutrients and
oxygen through the engineered tissues. Enough neovascularization can be attained by the proper use of angiogenic factors with appropriate cell types in scaold
biomaterials (Figure 1). There have been many eorts
to promote and regulate vascularization of engineered
tissues and also in pathological situations such as
chronic wounds in diabetic ulcers that are resulting
from insucient blood supply, contributing to inammation and infection at the decient sites,21 and myocardial ischemia, which is a weakening defect associated
with hypoxia and tissue necrosis because of occluded
vessels.171 Therefore, induction of neovascularization in

engineered tissues, chronic wounds, and ischemic areas


is the main therapeutic goal.
First of all, we should understand angiogenesis process. The process of angiogenesis follows from a complex cascade of events including ECs activation,
migration, and proliferation, their arrangement into
immature vessels, addition of mural cells (pericytes
and SMCs), and matrix deposition as the vessels
mature.21,137,172 The molecular mechanisms regulating
each of these stages are being described, and it is
obvious that dierent growth factors act at distinct
steps of neovascularization. For instance, bFGF and
VEGF, which are heparin binding growth factors,
contribute to the initiation of angiogenesis, and
induce endothelial cell proliferation and migration.
Platelet-derived growth factor (PDGF) is a mitogen
and chemotactic agent that recruits pericytes and
SMCs. Finally, TGF-b causes ECM deposition for stabilization of new vessels.137,173,174 However, VEGF and
its receptors constitute the key signaling system for
angiogenic activity in tissue formation.175 ECM proteins that participate in neovascularization are laminin,
collagen type I, and collagen type IV.167 So, it is important to properly deliver these signaling molecules
locally and temporally to obtain the desired biological
outcomes, while avoiding unfavorable side-eects.
Angiogenesis of engineered constructs is encouraged
ex vivo by biomaterial design, cell seeding, and culture
conditions. In vitro pre-vascularized scaolds would
then be transplanted in vivo and promoted to unite
with the host vascular network. This approach requires

Variouas approches in
construction of
vascularized thick tissues

1-Seeding of MSCs
or/and EPCs + other
differentiated cell
types or stem cells into
scaffold, then
transplantation into
damaged site of tissue.

2-First, in vitro
prevascularization of
scaffolds with ECs,
EPCs, or OECs, then
implantation of
scaffold into damaged
site of tissue followed
by subsequent in vivo
assembly of other cell
types.

3. Incorporating of the homing


inducing factors (i.e. chemokines
such as SDF1) or angiogenic
factors (such as VEGF, PDGF,
bFGF) into scaffold, then
transplantation of this scaffod
into the site of injury, leading to
angiogenesis along with tissue
repair due to homing of EPCs
and MSCs, and recruitment of
differentiated tissue residentcells from vicinity of injured site.

Figure 2. Various strategies that can be used to construct vascularized thick tissues.
Notes: EC, endothelial cell; OEC, outgrowth endothelial cell; SDF1, stromal cell-derived factor 1. For the meaning of other abbreviations, see Figure 1.

Downloaded from jba.sagepub.com at University of Liverpool on March 27, 2016

Naderi et al.

various biochemical signals inserted in the scaolds to


imitate normal microenvironment and would lead to
elevated angiogenic potential of the seeded cells.
Sometimes, the compatible scaolds are manipulated
with ECM proteins, ECM-derived peptides, and signaling proteins arranged in special micropatterns to direct
angiogenesis.95,176,177 Angiogenic factors are incorporated into scaold biomaterials and then transplanted
in vivo for the recruitment of EPCs followed by subsequent assembly of other cell types.178180 In another
approach, instead of angiogenic factors, one can incorporate chemokines (such as SDF1) into the scaolds to
induce homing of MSCs or/and EPCs from peripheral
blood to the site of implant and ultimately augment
angiogenesis.71,133,145
Taken together, in terms of cell sources and angiogenic factors, for successful engineering of vascularized
thick tissues, some fundamental guidelines must be considered, which are summarized here.
(1) The scaold must be compatible with ECs growth,
formation of capillaries, and construction of the
target tissue.181 The matrix can be coated with a
substance such as collagen, laminin or bronectin
that allows attachment and growth of ECs and
tissue-specic cells.
(2) Angiogenic growth factors are required for proliferation of ECs and better formation of blood vessels. Therefore, one can incorporate the source of
an angiogenic factor, with slow and sustainedrelease kinetics, into the bioengineered tissue
before implantation, so that enhanced new capillary
ingrowth from the hosts vascular plexus is achieved
after in vivo transplantation.25,182 As an alternative,
cells within the engineered tissue can be genetically
modied to secrete angiogenic factors.183,184
Release of these factors and chemokines (such as
SDF1) from the implanted site can recruit circulating EPCs or MSCs into the scaold and induce the
angiogenesis of the tissue.71,145,185
(3) For the augmentation of tissue vascularization,
ECs, EPCs, or OECs can be inserted into the bioengineered tissue. These cells can constitute
capillaries within the tissues in vitro and link to
the hosts vasculature systems in vivo.58 The combination of these cells with the incorporation of a
prolonged reservoir for angiogenic factor secretion
from the scaold would be more eective.186 It is
also possible to use MSCs for induction of angiogenesis in the scaold through their paracrine
eects.150
(4) The normal angiogenic process in the engineered
tissue must be developed so that a functional vascular network will be achieved. Excessive production of angiogenic factors may give rise to

deformed, non-functional vessels. On the other


hand, low concentration of these factors may
result in non-eective capillary density. Therefore,
the creation of new blood vessels should follow the
kinetics of normal development in the vasculature.174,187 A combination of concentrations and
various periods of exposure to dierent angiogenic
factors should be tested to nd the best conditions
for high-eciency angiogenesis. For this end, it is
necessary to design excellent DDSs for engineering
of thick tissues, and these systems are discussed in
the following section.

Drug delivery systems


Despite the fact that many biomaterials can supply
essential mechanical support and attachment sites,
they cannot direct changes in cellular phenotype as eciently as growth factors. Binding of growth factors to
various biomaterials appears to be a relatively simple
task.188 For example, biotinylated polymeric biomaterials can be easily coupled to growth factors by streptavidin.189 This technique has been used to conjugate
RGD to PLAPEG copolymers.190
Growth factors are released by cells for immediate
signaling or are embedded in the ECM and released in a
controlled manner. Sequestering of growth factors in
the ECM allows their stabilization and provides
physical cues for cells through spatial presentation.
Controlled release of factors from the ECM is coordinated by extracellular degradation. Generally, these
processes contribute to growth factor delivery that is
responsive and dynamic, changing in accordance with
specic cellular requirements and processes.191
Although systemic delivery of single proteins is technically simple, the succeeding distribution of them to
every place in the body and consequently their rapid
degradation result in unfavorable side eects and
toxicity, and also an inadequate local concentration
for the desired time frame.29 Polymeric systems can
be successfully tricked to deliver small doses of factors
at distinct release rates directly to target cells (Figure 1).
Polymeric delivery systems made up of various natural
and synthetic biomaterials can grant controlled growth
factor delivery by diverse mechanisms. Various types of
these materials have been applied for regulated release
of bFGF and VEGF, including alginate hydrogels,
PLG microspheres, and porous PLGA scaffolds.72,192,193 The release proles of biomolecules
from these carriers can be controlled by diusion, polymer degradation, the dose of the factors loaded in the
system, and the composition of the scaold.
Biodegradable polymer systems are used to deliver proteins30,31,194,195 or plasmid DNAs encoding the desired

Downloaded from jba.sagepub.com at University of Liverpool on March 27, 2016

10

Journal of Biomaterials Applications 0(0)

factors.196,197 However, there is necessity for growth


factor delivery systems that are able to deliver multiple
factors with distinct release kinetics, which is required
for driving normal tissue development.54,198
Growth factors can be incorporated into the scaffolds by two approaches. The rst approach involves
adding a lyophilized factor like VEGF to the polymer
particles prior to processing the polymer into a porous
scaold, which results in the factor being largely associated with the surface of the polymer. In this method,
VEGF is subjected to rapid release (e.g., days to weeks
in duration). The second approach involves pre-encapsulating of a factor like PDGF in PLG microspheres.
Therefore, fabricating scaolds from these particles
results in a more even distribution of factors throughout the polymer, with release kinetics controlled by
degradation rate of the polymer used to construct
microspheres. The two approaches may be combined
by mixing particulate polymers containing the rst
factor with microspheres containing a pre-encapsulated
second factor to deliver two growth factors with dierent release rates. The particulate and microsphere
PLGAs are then fused to form a homogeneous combined scaold with an open-pore structure.106
In a few circumstances, we need to deliver several
factors with distinct release kinetics in desired time
frames. Therefore, the future of bioactive materials is
the design of smart biomaterials that respond to their
environment with predetermined responses in which
release is initiated by microenvironmental cues.
Although the design of smart biomaterials is in its
early stages of development, the potential for engineering these biomaterials has been presented by many
studies.4,199,200 Various pathologic conditions can lead
to the increase of local temperature or acidity, or to the
activation of matrix metalloproteinases (MMPs), and
these microenvironmental conditions can be exploited
by smart biomaterials.200,201 MMPs, which cleave
specic amino acid sequences, act locally in ECM,
and are normally expressed at low levels in restful tissues. Thus, MMP-sensitive linkers could be used to
couple factors to biomaterials. Lutolf et al., developed
hydrogels with MMP-sensitive linkages between
polyethylene glycol chains entrapping BMP2. This
strategy allowed rapid bone formation in rats due to
proteolytic invasiveness of the gels and subsequent
release of BMP2.202
Successful repair and regeneration strategies will
require quantitative insight of tissue microenvironment
and can be engineered via designing biomaterials,
which provide quantitative adhesion, growth, or migration signals to direct cellular dierentiation pathways
including angiogenesis and vascular maturation.
Angiogenesis with biomaterial-based drug- and celldelivery systems have been reviewed by several

papers.25,203,204 Mimicking biological patterning may


be especially useful to control tissue development processes such as neovascularization, where unguided or
uncontrolled growth can lead to pathological eects
including tumor growth, metastasis, and deformed vessels. Techniques developed for microarray patterning
and microcontact printing, micromolding, laser photolithography, and micro-electro-mechanical systems
(microuidic devices) may be useful to form gradients
of growth factors within the scaolds. In addition to
biomaterial-based DDSs, these technical micropatterning approaches might be valuable in the generation of
complex networks of temporally and spatially
controlled growth factor delivery to mimic the microand nano-topographies of natural ECM, creating
complex tissue architectures in scaold materials and
to regulate angiogenesis.4,7,95,167

Conclusion
Advances in bioactive biomaterials and DDSs allow
not only controlled release, but also protection of
factors from degradation. Design of these advanced
biomaterials will require substantial basic biological
insight, since dose, timing, spatial range of growth
factors delivery, and also the conditions for environmentally controlled release will be highly specic for
each target tissue and disease. Materials can be
designed to be multifunctional and smart, in order to
provide sequential signals with dierent release kinetics
for individual factors. Thus, new rationally designed
biomaterials provide exact control of multiple growth
factors release with distinct gradient in response to a
specic niche. Identication and manipulation of biomaterials that support appropriate cellular attachment
and proliferation and induce optimal angiogenic signaling pathways are critical for engineering of thick
tissues.
Organized neovascularization in engineered tissues
may allow development of thick tissues with large
mass and complexity. To reach this goal, several key
issues must be considered; rst, since normal tissue
regeneration and development follow a specic series
of spatially and temporally coordinated signaling
events, biocompatible scaolding biomaterials must
be designed in a way to yield tissue constructions in a
precisely controlled manner. On the other hand, properties of each scaold should be specic for full tissue
engineering and regeneration including its angiogenesis.
For example, the matrix should have a high degree of
porosity to allow the penetration of blood vessels into
the implant. There are many biomaterials in use today,
in clinical settings, with reasonable biocompatibility;
however, performance of these biomaterials in conjunction with incorporated bioactive factors needs to be

Downloaded from jba.sagepub.com at University of Liverpool on March 27, 2016

Naderi et al.

11

addressed. Second, the use of better sources of vascular


cell progenitors (OEC-type EPCs, for example) in combination with other tissue-specic cell types may alleviate cell sourcing problem that can hinder industrial
scale-up of engineered thick tissues. The third issue is
the need for better comprehension of the biology
behind neovascularization. Understanding the normal
route of angiogenesis provides basic data on which we
can build and optimize normal growth and development of vessels within scaolds. The fourth important
factor is nding a way to optimize fabrication of scaffoldbiomolecule hybrids. For example, organization
of biomolecules and cells in biomaterials needs to be
optimized to mimic tissue complexity, and micro- and
nano-patterning. The fth issue involves drug loading
and delivery methods, release proles, and gradients,
which need to be examined to achieve maximum ecacy of growth factor release, similar to that of normal
tissue development. Finally, we need to integrate vascularized tissue constructs with functional cells of interest and also with hosts vasculature networks. To create
functional heart, muscle, lung, etc., the native cell types
or their precursors have to be either included or
recruited into the scaolds along with vascular cell
types. The resulting interactions among multiple cell
types have to be carefully examined so that functional
tissues are regenerated with complete network of blood
vessels. Further investigations are required to address
these critical issues for improvement and engineering of
clinically large tissues, which is an important goal in
tissue engineering.
References
1. Langer R. Selected advances in drug delivery and tissue
engineering. J Control Release 1999; 62: 711.
2. Peters A, Brey DM and Burdick JA. High-throughput and
combinatorial technologies for tissue engineering applications. Tissue Eng 2009; 15B: 225239.
3. Khademhosseini A, Ferreira L, Blumling J, Yeh J, Karp
JM, Fukuda J, et al. Co-culture of human embryonic stem
cells with murine embryonic fibroblasts on microwellpatterned substrates. Biomaterials 2006; 27: 59685977.
4. Anderson DG, Putnam D, Lavik EB, Mahmood TA and
Langer R. Biomaterial microarrays: rapid, microscale
screening of polymer-cell interaction. Biomaterials 2005;
26: 48924897.
5. Pernagallo S, Diaz-Mochon JJ and Bradley MA.
Cooperative polymer-DNA microarray approach to biomaterial investigation. Lab Chip 2009; 9: 397403.
6. Mei Y, Goldberg M and Anderson D. The development of
high-throughput screening approaches for stem cell engineering. Curr Opin Chem Biol 2007; 11: 388393.
7. Anderson DG, Levenberg S and Langer R. Nanoliter-scale
synthesis of arrayed biomaterials and application to
human embryonic stem cells. Nat Biotechnol 2004; 22:
863866.

8. Hook AL, Anderson DG, Langer R, Williams P, Davies


MC and Alexander MR. High throughput methods
applied in biomaterial development and discovery.
Biomaterials 2010; 31: 187198.
9. Patel ZS and Mikos AG. Angiogenesis with biomaterialbased drug- and cell-delivery systems. J Biomater Sci,
Polym Ed 2004; 15: 701726.
10. Neuss S, Apel C, Buttler P, Denecke B, Dhanasingh A,
Ding X, et al. Assessment of stem cell/biomaterial combinations for stem cell-based tissue engineering.
Biomaterials 2008; 29: 302313.
11. Dawson E, Mapili G, Erickson K, Taqvi S and Roy K.
Biomaterials for stem cell differentiation. Adv Drug Deliv
Rev 2008; 60: 215228.
12. Beckstead BL, Santosa DM and Giachelli CM.
Mimicking cell-cell interactions at the biomaterial-cell
interface for control of stem cell differentiation. J
Biomed Mater Res 2006; 79A: 94103.
13. Mao C, Qiu Y, Sang H, Mei H, Zhu A, Shen J, et al.
Various approaches to modify biomaterial surfaces for
improving hemocompatibility. Adv Colloid Interface Sci
2004; 110: 517.
14. Ahsan T and Nerem RM. Bioengineered tissues: the science, the technology, and the industry. Orthod Craniofac
Res 2005; 8: 134140.
15. Yarlagadda PK, Chandrasekharan M and Shyan JY.
Recent Advances and current developments in tissue scaffolding. Biomed Mater Eng 2005; 15: 159177.
16. Fisher OZ, Khademhosseini A, Langer R and Peppas
NA. Bioinspired materials for controlling stem cell fate.
Acc Chem Res 2010; 43: 419428.
17. Yoshikawa H and Myoui A. Bone tissue engineering with
porous hydroxyapatite ceramics. J Artif Organs 2005; 8:
131136.
18. Lacroix D, Planell JA and Prendergast PJ. Computeraided design and finite-element modelling of biomaterial
scaffolds for bone tissue engineering. Philos Transact A
Math Phys Eng Sci 2009; 367: 19932009.
19. Raghunath J, Salacinski HJ, Sales KM, Butler PE and
Seifalian AM. Advancing cartilage tissue engineering: the
application of stem cell technology. Curr Opin Biotechnol
2005; 16: 503509.
20. Chin CD, Khanna K and Sia SK. A microfabricated
porous collagen-based scaffold as prototype for skin substitutes. Biomed Microdevices 2008; 10: 459467.
21. Nillesen ST, Geutjes PJ, Wismans R, Schalkwijk J,
Daamen WF and van Kuppevelt TH. Increased angiogenesis and blood vessel maturation in acellular collagenheparin scaffolds containing both FGF2 and VEGF.
Biomaterials 2007; 28: 11231131.
22. Elisseeff J, Puleo C, Yang F and Sharma B. Advances in
skeletal tissue engineering with hydrogels. Orthod
Craniofac Res 2005; 8: 150161.
23. Tabata Y. Biomaterial technology for tissue engineering
applications. J R Soc Interface 2009; 6: S311S324.
24. Unger RE, Sartoris A, Peters K, Motta A, Migliaresi C,
Kunkel M, et al. Tissue-like self-assembly in cocultures of
endothelial cells and osteoblasts and the formation
of microcapillary-like structures on three-dimensional
porous biomaterials. Biomaterials 2007; 28: 39653976.

Downloaded from jba.sagepub.com at University of Liverpool on March 27, 2016

12

Journal of Biomaterials Applications 0(0)

25. Laschke MW, Harder Y, Amon M, Martin I, Farhadi J,


Ring A, et al. Angiogenesis in tissue engineering: breathing life into constructed tissue substitutes. Tissue Eng
2006; 12: 20932104.
26. Ko HC, Milthorpe BK and McFarland CD. Engineering
thick tissues: the vascularisation problem. Eur Cell Mater
2007; 14: 118.
27. Cleland JL, Duenas ET, Park A, Daugherty A, Kahn J,
Kowalski J, et al. Development of poly-(D, L-lactide-coglycolide) microsphere formulations containing recombinant human vascular endothelial growth factor to
promote local angiogenesis. J Control Release 2001; 72:
1324.
28. Lee J and Lee KY. Local and sustained vascular endothelial growth factor delivery for angiogenesis using an
injectable system. Pharm Res 2009; 26: 17391744.
29. Silva EA and Mooney DJ. Spatiotemporal control of
vascular endothelial growth factor delivery from injectable hydrogels enhances angiogenesis. J Thromb Haemost
2007; 5: 590598.
30. Langer R and Tirrell DA. Designing materials for biology and medicine. Nature 2004; 428: 487492.
31. Kohane DS and Langer R. Polymeric biomaterials in
tissue engineering. Pediatr Res 2008; 63: 487491.
32. Lovett M, Lee K, Edwards A and Kaplan DL.
Vascularization strategies for tissue engineering. Tissue
Eng 2009; 15B: 353370.
33. Davis ME, Hsieh PC, Grodzinsky AJ and Lee RT.
Custom design of the cardiac microenvironment with biomaterials. Circ Res 2005; 97: 815.
34. Sundelacruz S and Kaplan DL. Stem cell- and scaffoldbased tissue engineering approaches to osteochondral
regenerative medicine. Semin Cell Dev Biol 2009; 20:
646655.
35. Hutmacher DW and Cool S. Concepts of scaffold-based
tissue engineering: the rationale to use solid free-form fabrication techniques. J Cell Mol Med 2007; 11: 654669.
36. Garreta E, Gasset D, Semino C and Borros S.
Fabrication of a three-dimensional nanostructured biomaterial for tissue engineering of bone. Biomol Eng
2007; 24: 7580.
37. Berthold A, Haibel A, Brandes N, Kroh L, Gross U,
Uharek L, et al. Biocompatible porous ceramics for the
cultivation of hematopoietic cells. J Mater Sci Mater
Med 2007; 18: 13331338.
38. Willerth SM, Arendas KJ, Gottlieb DI and SakiyamaElbert SE. Optimization of fibrin scaffolds for differentiation of murine embryonic stem cells into neural lineage
cells. Biomaterials 2006; 27: 59906003.
39. Willerth SM and Sakiyama-Elbert SE. Approaches
to neural tissue engineering using scaffolds for drug delivery. Adv Drug Deliv Rev 2007; 59: 325338.
40. Cheng NC, Estes BT, Awad HA and Guilak F.
Chondrogenic differentiation of adipose-derived adult
stem cells by a porous scaffold derived from native articular cartilage extracellular matrix. Tissue Eng 2009; 15A:
231241.
41. Burdick JA and Vunjak-Novakovic G. Engineered
microenvironments for controlled stem cell differentiation. Tissue Eng 2009; 15A: 205219.

42. Shin M, Abukawa H, Troulis MJ and Vacanti JP.


Development of a biodegradable scaffold with interconnected pores by heat fusion and its application to bone
tissue engineering. J Biomed Mater Res 2008; 84A: 702709.
43. Patel M and Fisher JP. Biomaterial scaffolds in pediatric
tissue engineering. Pediatr Res 2008; 63: 497501.
44. Jeong SI, Kim BS, Lee YM, Ihn KJ, Kim SH and
Kim YH. Morphology of elastic poly(L-lactide-coepsilon-caprolactone) copolymers and in vitro and
in vivo degradation behavior of their scaffolds. Biomacromolecules 2004; 5: 13031309.
45. Jeong SI, Kim BS, Kang SW, Kwon JH, Lee YM, Kim SH,
et al. In vivo biocompatibilty and degradation behavior
of elastic poly(L-lactide-co-epsilon-caprolactone) scaffolds. Biomaterials 2004; 25: 59395946.
46. Hayami JW, Surrao DC, Waldman SD and Amsden BG.
Design and characterization of a biodegradable composite scaffold for ligament tissue engineering. J Biomed
Mater Res 2010; 92A: 14071420.
47. Kong HJ, Kaigler D, Kim K and Mooney DJ.
Controlling rigidity and degradation of alginate hydrogels via molecular weight distribution. Biomacromolecules
2004; 5: 17201727.
48. Salem AK, Stevens R, Pearson RG, Davies MC,
Tendler SJ, Roberts CJ, et al. Interactions of 3T3 fibroblasts and endothelial cells with defined pore features.
J Biomed Mater Res 2002; 61: 212217.
49. Guan J, Stankus JJ and Wagner WR. Biodegradable elastomeric scaffolds with basic fibroblast growth factor
release. J Control Release 2007; 120: 7078.
50. Wei G, Jin Q, Giannobile WV and Ma PX. Nano-fibrous
scaffold for controlled delivery of recombinant human
PDGF-BB. J Control Release 2006; 112: 103110.
51. Botchwey EA, Dupree MA, Pollack SR, Levine EM and
Laurencin CT. Tissue engineered bone: measurement of
nutrient transport in three-dimensional matrices. J
Biomed Mater Res 2003; 67A: 357367.
52. Narmoneva DA, Oni O, Sieminski AL, Zhang S, Gertler
JP, Kamm RD, et al. Self-assembling short oligopeptides
and the promotion of angiogenesis. Biomaterials 2005; 26:
48374846.
53. Zawaneh PN, Singh SP, Padera RF, Henderson PW,
Spector JA and Putnam D. Design of an injectable synthetic and biodegradable surgical biomaterial. Proc Nat
Acad Sci USA 2010; 107: 1101411019.
54. Sun Q, Silva EA, Wang A, Fritton JC, Mooney DJ,
Schaffler MB, et al. Sustained release of multiple
growth factors from injectable polymeric system as a
novel therapeutic approach towards angiogenesis.
Pharm Res 2010; 27: 264271.
55. Wang HB, Zhou J, Liu ZQ and Wang CY. Injectable
cardiac tissue engineering for the treatment of myocardial
infarction. J Cell Mol Med 2010; 14: 10441055.
56. Nguyen MK and Lee DS. Injectable biodegradable
hydrogels. Macromol Biosci 2010; 10: 563579.
57. Nolan K, Millet Y, Ricordi C and Stabler CL. Tissue
engineering and biomaterials in regenerative medicine.
Cell Transplant 2008; 17: 241243.
58. Fuchs S, Motta A, Migliaresi C and Kirkpatrick CJ.
Outgrowth endothelial cells isolated and expanded from

Downloaded from jba.sagepub.com at University of Liverpool on March 27, 2016

Naderi et al.

59.

60.

61.

62.

63.

64.

65.

66.

67.

68.

69.

70.

71.

72.

13

human peripheral blood progenitor cells as a potential


source of autologous cells for endothelialization of silk
fibroin biomaterials. Biomaterials 2006; 27: 53995408.
Kogan G, Soltes L, Stern R and Gemeiner P. Hyaluronic
acid: a natural biopolymer with a broad range of biomedical and industrial applications. Biotechnol Lett 2007; 29:
1725.
Silva GA, Costa FJ, Neves NM and Reis RL.
Microparticulate release systems based on natural origin
materials. Adv Exp Med Biol 2004; 553: 283300.
Eiselt P, Yeh J, Latvala RK, Shea LD and Mooney DJ.
Porous carriers for biomedical applications based on alginate hydrogels. Biomaterials 2000; 21: 19211927.
Shi CM, Zhu Y, Ran XZ, Wang M, Su YP and Cheng
TM. Therapeutic potential of chitosan and its derivatives in regenerative medicine. J Surg Res 2006; 133:
185192.
Kleinman HK, McGarvey ML, Liotta LA, Robey PG,
Tryggvason K and Martin GR. Isolation and characterization of type IV procollagen, laminin, and heparan sulfate proteoglycan from the EHS sarcoma. Biochemistry
1982; 21: 61886193.
Laschke MW, Rucker M, Jensen G, Carvalho C,
Mulhaupt R, Gellrich NC, et al. Incorporation of
growth factor containing matrigel promotes vascularization of porous PLGA scaffolds. J Biomed Mater Res
2008; 85A: 397407.
Melero-Martin JM, Khan ZA, Picard A, Wu X,
Paruchuri S and Bischoff J. In vivo vasculogenic potential
of human blood-derived endothelial progenitor cells.
Blood 2007; 109: 47614768.
Brun P, Abatangelo G, Radice M, Zacchi V, Guidolin D,
Gordini DD, et al. Chondrocyte aggregation and reorganization into three-dimensional scaffolds. J Biomed
Mater Res 1999; 46: 337346.
Gerecht S, Burdick JA, Ferreira LS, Townsend SA,
Langer R and Vunjak-Novakovic G. Hyaluronic acid
hydrogel for controlled self-renewal and differentiation
of human embryonic stem cells. Proc Nat Acad Sci
USA 2007; 104: 1129811303.
Zhao H, Ma L, Zhou J, Mao Z, Gao C and Shen J.
Fabrication and physical and biological properties of
fibrin gel derived from human plasma. Biomed Mater
2008; 3: 15001.
Westreich R, Kaufman M, Gannon P and Lawson W.
Validating the subcutaneous model of injectable autologous cartilage using a fibrin glue scaffold. Laryngoscope
2004; 114: 21542160.
Christman KL, Vardanian AJ, Fang Q, Sievers RE, Fok
HH and Lee RJ. Injectable fibrin scaffold improves cell
transplant survival, reduces infarct expansion, and
induces neovasculature formation in ischemic myocardium. J Am Coll Cardiol 2004; 44: 654660.
Thevenot PT, Nair AM, Shen J, Lotfi P, Ko CY and
Tang L. The effect of incorporation of SDF-1alpha into
PLGA scaffolds on stem cell recruitment and the inflammatory response. Biomaterials 2010; 31: 39974008.
Wang Y, Liu XC, Zhao J, Kong XR, Shi RF, Zhao
XB, et al. Degradable PLGA scaffolds with basic
fibroblast growth factor experimental studies in

73.

74.

75.

76.

77.

78.

79.

80.

81.

82.

83.

84.

85.

86.

87.

myocardial revascularization. Tex Heart Inst J 2009;


36: 8997.
Young CS, Abukawa H, Asrican R, Ravens M, Troulis
MJ, Kaban LB, et al. Tissue-engineered hybrid tooth and
bone. Tissue Eng 2005; 11: 15991610.
Cima LG, Vacanti JP, Vacanti C, Ingber D, Mooney D
and Langer R. Tissue engineering by cell transplantation
using degradable polymer substrates. J Biomech Eng
1991; 113: 143151.
Mondrinos MJ, Koutzaki SH, Poblete HM, Crisanti MC,
Lelkes PI and Finck CM. In vivo pulmonary tissue engineering: contribution of donor-derived endothelial cells
to construct vascularization. Tissue Eng 2008; 14:
361368.
Borden M, Attawia M and Laurencin CT. The sintered
microsphere matrix for bone tissue engineering: in vitro
osteoconductivity studies. J Biomed Mater Res 2002; 61:
421429.
Chai C and Leong KW. Biomaterials approach to expand
and direct differentiation of stem cells. Mol Ther 2007; 15:
467480.
Nitzsche H, Lochmann A, Metz H, Hauser A, Syrowatka
F, Hempel E, et al. Fabrication and characterization of a
biomimetic composite scaffold for bone defect repair.
J Biomed Mater Res 2010; 94A: 298307.
Kim HW, Knowles JC and Kim HE. Porous scaffolds of
gelatin-hydroxyapatite nanocomposites obtained by biomimetic approach: characterization and antibiotic drug
release. J Biomed Mater Res Appl Biomater 2005; 74B:
686698.
Levenberg S, Huang NF, Lavik E, Rogers AB, ItskovitzEldor J and Langer R. Differentiation of human embryonic stem cells on three-dimensional polymer scaffolds.
Proc Nat Acad Sci USA 2003; 100: 1274112746.
Luna SM, Silva SS, Gomez ME, Mano JF and Reis RL.
Cell adhesion and proliferation onto chitosan-based
membranes treated by plasma surface modification.
J Biomater Appl 2010 [Epub ahead of print].
Zippel N, Schulze M and Tobiasch E. Biomaterials and
mesenchymal stem cells for regenerative medicine. Recent
Pat Biotechnol 2010; 4: 122.
Curran JM, Chen R and Hunt JA. The guidance of
human mesenchymal stem cell differentiation in vitro by
controlled modifications to the cell substrate.
Biomaterials 2006; 27: 47834793.
Blan NR and Birla RK. Design and fabrication of heart
muscle using scaffold-based tissue engineering. J Biomed
Mater Res 2008; 86A: 195208.
Holtorf HL, Jansen JA and Mikos AG. Ectopic bone
formation in rat marrow stromal cell/titanium fiber
mesh scaffold constructs: effect of initial cell phenotype.
Biomaterials 2005; 26: 62086216.
Nuttelman CR, Tripodi MC and Anseth KS. In vitro
osteogenic differentiation of human mesenchymal stem
cells photoencapsulated in PEG hydrogels. J Biomed
Mater Res 2004; 68A: 773782.
Li YJ, Chung EH, Rodriguez RT, Firpo MT and Healy
KE. Hydrogels as artificial matrices for human embryonic stem cell self-renewal. J Biomed Mater Res 2006;
79A: 15.

Downloaded from jba.sagepub.com at University of Liverpool on March 27, 2016

14

Journal of Biomaterials Applications 0(0)

88. Park YD, Tirelli N and Hubbell JA. Photopolymerized


hyaluronic acid-based hydrogels and interpenetrating
networks. Biomaterials 2003; 24: 893900.
89. Mann BK, Gobin AS, Tsai AT, Schmedlen RH and
West JL. Smooth muscle cell growth in photopolymerized hydrogels with cell adhesive and proteolytically
degradable domains: synthetic ECM analogs for tissue
engineering. Biomaterials 2001; 22: 30453051.
90. Hsiong SX, Huebsch N, Fischbach C, Kong HJ and
Mooney DJ. Integrin-adhesion ligand bond formation
of preosteoblasts and stem cells in three-dimensional
RGD presenting matrices. Biomacromolecules 2008; 9:
18431851.
91. Patel PN, Gobin AS, West JL and Patrick CW.
Poly(ethylene glycol) hydrogel system supports preadipocyte viability, adhesion, and proliferation. Tissue Eng
2005; 11: 14981505.
92. Oliveira JM, Sousa RA, Kotobuki N, Tadokoro M,
Hirose M, Mano JF, et al. The osteogenic differentiation of rat bone marrow stromal cells cultured
with dexamethasone-loaded carboxymethylchitosan/
poly(amidoamine)
dendrimer
nanoparticles.
Biomaterials 2009; 30: 804813.
93. Baier Leach J, Bivens KA, Patrick CW and Schmidt CE.
Photocrosslinked hyaluronic acid hydrogels: natural,
biodegradable tissue engineering scaffolds. Biotechnol
Bioeng 2003; 82: 578589.
94. Hatakeyama H, Kikuchi A, Yamato M and Okano T.
Bio-functionalized thermoresponsive interfaces facilitating cell adhesion and proliferation. Biomaterials 2006;
27: 50695078.
95. Moon JJ, Hahn MS, Kim I, Nsiah BA and West JL.
Micropatterning of poly(ethylene glycol) diacrylate
hydrogels with biomolecules to regulate and guide endothelial morphogenesis. Tissue Eng 2009; 15A: 579585.
96. Jun HW and West JL. Modification of polyurethaneurea with PEG and YIGSR peptide to enhance
endothelialization without platelet adhesion. J Biomed
Mater Res Appl Biomater 2005; 72B: 131139.
97. Kouvroukoglou S, Dee KC, Bizios R, McIntire LV and
Zygourakis K. Endothelial cell migration on surfaces modified with immobilized adhesive peptides.
Biomaterials 2000; 21: 17251733.
98. Fittkau MH, Zilla P, Bezuidenhout D, Lutolf M, Human
P, Hubbell JA, et al. The selective modulation of endothelial cell mobility on RGD peptide containing surfaces by
YIGSR peptides. Biomaterials 2005; 26: 167174.
99. Massia SP and Hubbell JA. Vascular endothelial-cell
adhesion and spreading promoted by the peptide
RGDY of the IIICS region of plasma fibronectin is
mediated by integrin alpha-4-beta-1. J Biol Chem 1992;
267: 1401914026.
100. Heilshorn SC, DiZio KA, Welsh ER and Tirrell DA.
Endothelial cell adhesion to the fibronectin CS5
domain in artificial extracellular matrix proteins.
Biomaterials 2003; 24: 42454252.
101. Lu Y, Mapili G, Suhali G, Chen S and Roy K. A digital
micro-mirror device-based system for the microfabrication of complex, spatially patterned tissue engineering
scaffolds. J Biomed Mater Res 2006; 77A: 396405.

102. Mapili G, Lu Y, Chen S and Roy K. Laser-layered


microfabrication of spatially patterned functionalized
tissue-engineering scaffolds. J Biomed Mater Appl
Biomater 2005; 75B: 414424.
103. Tsutsumi S, Shimazu A, Miyazaki K, Pan H, Koike C,
Yoshida E, et al. Retention of multilineage differentiation potential of mesenchymal cells during proliferation
in response to FGF. Biochem Biophys Res Commun
2001; 288: 413419.
104. Reddi AH. Morphogenesis and tissue engineering of
bone and cartilage: inductive signals, stem cells, and biomimetic biomaterials. Tissue Eng 2000; 6: 351359.
105. Jansen JA, Vehof JWM, Ruhe PQ, Kroeze-Deutman H,
Kuboki Y, Takita H, et al. Growth factor-loaded scaffolds for bone engineering. J Control Release 2005; 101:
127136.
106. Richardson TP, Peters MC, Ennett AB and Mooney DJ.
Polymeric system for dual growth factor delivery. Nat.
Biotechnol 2001; 19: 10291034.
107. Koch S, Yao C, Grieb G, Prevel P, Noah EM and
Steffens GC. Enhancing angiogenesis in collagen matrices by covalent incorporation of VEGF. J Mater Sci
Mater Med 2006; 17: 735741.
108. Steffens GC, Yao C, Prevel P, Markowicz M, Schenck
P, Noah EM, et al. Modulation of angiogenic potential
of collagen matrices by covalent incorporation of heparin and loading with vascular endothelial growth factor.
Tissue Eng 2004; 10: 15021509.
109. Zisch AH, Schenk U, Schense JC, Sakiyama-Elbert SE
and Hubbell JA. Covalently conjugated VEGF-fibrin
matrices for endothelialization. J Control Release 2001;
72: 101113.
110. DeLong SA, Moon JJ and West JL. Covalently immobilized gradients of bFGF on hydrogel scaffolds for directed cell migration. Biomaterials 2005; 26: 32273234.
111. Zisch AH, Zeisberger SM, Ehrbar M, Djonov V, Weber
CC, Ziemiecki A, et al. Engineered fibrin matrices for
functional display of cell membrane-bound growth
factor-like activities: study of angiogenic signaling by
ephrin-B2. Biomaterials 2004; 25: 32453257.
112. Moon JJ, Lee SH and West JL. Synthetic biomimetic
hydrogels incorporated with ephrin-A1 for therapeutic
angiogenesis. Biomacromolecules 2007; 8: 4249.
113. Czyz J, Wiese C, Rolletschek A, Blyszczuk P, Cross M
and Wobus AM. Potential of embryonic and adult stem
cells in vitro. J Biol Chem 2003; 384: 13911409.
114. Harun R, Ruban L, Matin M, Draper J, Jenkins N,
Liew G, et al. Cytotrophoblast stem cell lines derived
from human embryonic stem cells and their capacity to
mimic invasive implantation events. Hum Reprod 2006;
21: 13491358.
115. Wakitani S, Takaoka K, Hattori T, Miyazawa N,
Iwanaga T, Takeda S, et al. Embryonic stem cells
injected into the mouse knee joint form teratomas and
subsequently destroy the joint. Rheumatology 2003; 42:
162165.
116. Andrews PW, Matin MM, Bahrami AR, Damjanov I,
Gokhale P and Draper JS. Embryonic stem (ES) cells
and embryonal carcinoma (EC) cells: opposite sides of
the same coin. Biochem Soc Trans 2005; 33: 15261530.

Downloaded from jba.sagepub.com at University of Liverpool on March 27, 2016

Naderi et al.

15

117. Polak JM and Bishop AE. Stem cells and tissue engineering: past, present, and future. Ann NY Acad Sci
2006; 1068: 352366.
118. Enver T, Soneji S, Joshi C, Brown J, Iborra F, Orntoft
T, et al. Cellular differentiation hierarchies in normal
and culture-adapted human embryonic stem cells. Hum
Mol Genet 2005; 14: 31293140.
119. Draper JS, Smith K, Gokhale P, Moore HD, Maltby E,
Johnson J, et al. Recurrent gain of chromosomes 17q
and 12 in cultured human embryonic stem cells. Nat
Biotechnol 2004; 22: 5354.
120. Zwaginga JJ and Doevendans P. Stem cell-derived
angiogenic/vasculogenic cells: possible therapies for
tissue repair and tissue engineering. Clin Exp Pharm
Physiol 2003; 30: 900908.
121. Sakai T, Ling YQ, Payne TR and Huard J. The use of
Ex Vivo gene transfer based on muscle-derived stem cells
for cardiovascular medicine. Trends Cardiov Med 2002;
12: 115120.
122. Gronthos S, Zannettino ACW, Hay SJ, Shi ST, Graves
SE, Kortesidis A, et al. Molecular and cellular characterisation of highly purified stromal stem cells derived
from human bone marrow. J Cell Sci 2003; 116:
18271835.
123. Rezai N, Podor TJ and McManus BM. Bone marrow
cells in the repair and modulation of heart and blood
vessels: emerging opportunities in native and engineered
tissue and biomechanical materials. Artif Organs 2004;
28: 142151.
124. Amin EM, Reza BA, Morteza BR, Maryam MM, Ali M
and Zeinab N. Microanatomical evidences for potential
of mesenchymal stem cells in amelioration of striatal
degeneration. Neurol Res 2008; 30: 10861090.
125. Edalatmanesh MA, Matin MM, Neshati Z, Bahrami
AR and Kheirabadi M. Systemic transplantation of
mesenchymal stem cells can reduce cognitive and
motor deficits in rats with unilateral lesions of the neostriatum. Neurol Res 2010; 32: 166172.
126. Neshati Z, Matin MM, Bahrami AR and Moghimi A.
Differentiation of mesenchymal stem cells to insulinproducing cells and their impact on type 1 diabetic
rats. J Physiol Biochem 2010; 66: 181187.
127. Hoffmann J, Glassford AJ, Doyle TC, Robbins RC,
Schrepfer S and Pelletier MP. Angiogenic effects despite
limited cell survival of bone marrow-derived mesenchymal stem cells under ischemia. Thorac Cardiovasc Surg
2010; 58: 136142.
128. Kim SY and Im GI. Updates on scaffold application
and vascularization in bone tissue engineering. Tissue
Eng Reg Med 2009; 6: 13911400.
129. Gnecchi M and Melo LG. Bone marrow-derived mesenchymal stem cells: isolation, expansion, characterization, viral transduction, and production of conditioned
medium. Methods Mol Biol 2009; 482: 281294.
130. Mihu CM, Mihu D, Costin N, Ciuca DR, Susman S and
Ciortea R. Isolation and characterization of stem cells
from the placenta and the umbilical cord. Rom J
Morphol Embryol 2008; 49: 441446.
131. Patel AN, Park E, Kuzman M, Benetti F, Silva FJ and
Allickson JG. Multipotent menstrual blood stromal

132.

133.

134.

135.

136.
137.

138.

139.

140.

141.

142.

143.

144.

145.

146.

stem cells: isolation, characterization, and differentiation. Cell Transplant 2008; 17: 303311.
Riekstina U, Cakstina I, Muceniece R, Jankovskis G
and Ancans J. Isolation, cultivation and characterization of human somatic stem cells from adult skin,
adipose tissue and bone marrow. Cell Res 2008; 18:
S161.
Chavakis E, Urbich C and Dimmeler S. Homing and
engraftment of progenitor cells: a prerequisite for cell
therapy. J Mol Cell Cardiol 2008; 45: 514522.
Ahmadian Kia N, Bahrami AR, Ebrahimi M, Matin
MM, Neshati Z, Almohaddesin MR, et al.
Comparative analysis of chemokine receptors expression in mesenchymal stem cells derived from human
bone marrow and adipose tissue. J Mol Neurosci 2010
[Epub ahead of print].
Ghadge SK, Muhlstedt S, Ozcelik C and Bader M.
SDF-1alpha as a therapeutic stem cell homing factor
in myocardial infarction. Pharm Ther 2011; 129: 97108.
Karamysheva AF. Mechanisms of angiogenesis.
Biochemistry (Mosc) 2008; 73: 751762.
Silvestre JS, Levy BI and Tedgui A. Mechanisms of
angiogenesis and remodelling of the microvasculature.
Cardiovasc Res 2008; 78: 201202.
Pepper MS, Mandriota SJ, Vassalli JD, Orci L and
Montesano R. Angiogenesis-regulating cytokines: activities and interactions. Curr Top Microbiol Immunol 1996;
213: 3167.
Asahara T, Murohara T, Sullivan A, Silver M, van der
Zee R, Li T, et al. Isolation of putative progenitor endothelial cells for angiogenesis. Science 1997; 275: 964967.
Asahara T, Takahashi T, Masuda H, Kalka C, Chen
DH, Iwaguro H, et al. VEGF contributes to postnatal
neovascularization by mobilizing bone marrow-derived
endothelial progenitor cells. EMBO J 1999; 18:
39643972.
Wijelath ES, Rahman S, Murray J, Patel Y, Savidge G
and Sobel M. Fibronectin promotes VEGF-induced
CD34() cell differentiation into endothelial cells. J
Vasc Surg 2004; 39: 655660.
Lu H, Feng Z, Gu Z and Liu C. Growth of outgrowth
endothelial cells on aligned PLLA nanofibrous scaffolds. J Mater Sci Mater Med 2009; 20: 19371944.
Gulati R, Jevremovic D, Peterson TE, Chatterjee S,
Shah V, Vile RG and Simari RD. Diverse origin and
function of cells with endothelial phenotype obtained
from adult human blood. Circ Res 2003; 93: 10231025.
Matsumura G, Miyagawa-Tomita S, Shinoka T, Ikada
Y and Kurosawa H. First evidence that bone marrow
cells contribute to the construction of tissue-engineered
vascular autografts in vivo. Circulation 2003; 108:
17291734.
Hoenig MR, Bianchi C and Sellke FW. Hypoxia inducible factor-1 alpha, endothelial progenitor cells, monocytes, cardiovascular risk, wound healing, cobalt and
hydralazine: a unifying hypothesis. Curr Drug Targets
2008; 9: 422435.
Hur J, Yoon CH, Kim HS, Choi JH, Kang HJ, Hwang
KK, et al. Characterization of two types of endothelial
progenitor cells and their different contributions to

Downloaded from jba.sagepub.com at University of Liverpool on March 27, 2016

16

147.

148.

149.

150.

151.

152.

153.

154.

155.

156.

157.

158.

Journal of Biomaterials Applications 0(0)


neovasculogenesis. Arterioscler Thromb Vasc Biol 2004;
24: 288293.
Kinnaird T, Stabile E, Burnett MS, Lee CW, Barr S,
Fuchs S, et al. Marrow-derived stromal cells express
genes encoding a broad spectrum of arteriogenic cytokines and promote in vitro and in vivo arteriogenesis
through paracrine mechanisms. Circ Res 2004; 94:
678685.
Jin DK, Shido K, Kopp HG, Petit I, Shmelkov SV,
Young LM, et al. Cytokine-mediated deployment of
SDF-1 induces revascularization through recruitment of
CXCR4() hemangiocytes. Nat Med 2006; 12: 557567.
Zhang J, Zhang XZ and Zhang Y. Role of mesenchymal
stem cells in angiogenesis and clinical applications.
Zhongguo Shi Yan Xue Ye Xue Za Zhi 2010; 18:
10841087.
Ahmadi R, Burns AJ and de Bruijn JD. Chitosan-based
hydrogels do not induce angiogenesis. J Tissue Eng
Regen Med 2010; 4: 309315.
Yoon CH, Hur J, Park KW, Kim JH, Lee CS, Oh IY,
et al. Synergistic neovascularization by mixed transplantation of early endothelial progenitor cells and late outgrowth endothelial cells: the role of angiogenic cytokines
and matrix metalloproteinases. Circulation 2005; 112:
16181627.
Ziegelhoeffer T, Fernandez B, Kostin S, Heil M,
Nauheim B, Voswinckel R, et al. Bone marrow-derived
cells do not incorporate in the adult growing vasculature. Circ Res 2004; 94: 230238.
Rookmaaker MB, Vergeer M, van Zonneveld AJ,
Rabelink TJ and Verhaar MC. Endothelial progenitor
cells: mainly derived from the monocytemacrophagecontaining CD34(-) mononuclear cell population and
only in part from the hematopoietic stem cell-containing
CD34() mononuclear cell population. Circulation
2003; 107: 11641169.
Fujiyama S, Matsubara H, Amano K, Iba O, Shibasaki
Y, Okigaki M, et al. Bone marrow monocyte-lineage
cells cause reendothelialization as endothelial progenitors in vascular repair in MCP-1-dependent manner.
Circulation 2002; 106: 82.
Masuda H and Asahara T. Post-natal endothelial progenitor cells for neovascularization in tissue regeneration. Cardiovasc Res 2003; 58: 390398.
Dvorin EL, Wylie-Sears J, Kaushal S, Martin DP and
Bischoff J. Quantitative evaluation of endothelial progenitors and cardiac valve endothelial cells: proliferation
and differentiation on poly-glycolic acid/poly-4-hydroxybutyrate scaffold in response to vascular endothelial
growth factor and transforming growth factor beta 1.
Tissue Eng 2003; 9: 487493.
Zhu C, Ying D, Zhou D, Mi J, Zhang W, Chang Q,
et al. Expression of TGF-beta1 in smooth muscle cells
regulates endothelial progenitor cells migration and
differentiation. J Surg Res 2005; 125: 151156.
Badorff C, Brandes RP, Popp R, Rupp S, Urbich C,
Aicher A, et al. Transdifferentiation of blood-derived
human adult endothelial progenitor cells into functionally active cardiomyocytes. Circulation 2003; 107:
10241032.

159. Schumann P, Tavassol F, Lindhorst D, Stuehmer C,


Bormann KH, Kampmann A, et al. Consequences of
seeded cell type on vascularization of tissue engineering
constructs in vivo. Microvasc Res 2009; 78: 180190.
160. Fuchs S, Ghanaati S, Orth C, Barbeck M, Kolbe M,
Hofmann A, et al. Contribution of outgrowth endothelial cells from human peripheral blood on in vivo vascularization of bone tissue engineered constructs based on
starch polycaprolactone scaffolds. Biomaterials 2009;
30: 526534.
161. Unger RE, Sartoris A, Peters K, Motta A, Migliaresi C,
Kunkel M, et al. Tissue-like self-assembly in cocultures
of endothelial cells and osteoblasts and the formation of
microcapillary-like structures on three-dimensional
porous biomaterials. Biomaterials 2007; 28: 39653976.
162. Nomi M, Atala A, Coppi PD and Soker S. Principals of
neovascularization for tissue engineering. Mol Aspects
Med 2002; 23: 463483.
163. Co CC, Wang YC and Ho CC. Biocompatible micropatterning of two different cell types. J Am Chem Soc
2005; 127: 15981599.
164. Risbud MV, Karamuk E, Moser R and Mayer J.
Hydrogel-coated textile scaffolds as three-dimensional
growth support for human umbilical vein endothelial
cells (HUVECs): possibilities as coculture system in
liver tissue engineering. Cell Transplant 2002; 11:
369377.
165. Hwang NS, Varghese S and Elisseeff J. Controlled differentiation of stem cells. Adv Drug Deliv Rev 2008; 60:
199214.
166. Dawson JI and Oreffo RO. Bridging the regeneration
gap: stem cells, biomaterials and clinical translation in
bone tissue engineering. Arch Biochem Biophys 2008;
473: 124131.
167. Moon JJ and West JL. Vascularization of engineered
tissues: approaches to promote angiogenesis in biomaterials. Curr Top Med Chem 2008; 8: 300310.
168. Eiselt P, Kim BS, Chacko B, Isenberg B, Peters MC,
Greene KG, et al. Development of technologies aiding
large-tissue engineering. Biotechnol Prog 1998; 14:
134140.
169. Atala A. Advances in tissue and organ replacement.
Curr Stem Cell Res Ther 2008; 3: 2131.
170. Naughton GK. From lab bench to market: critical issues
in tissue engineering. Ann NY Acad Sci 2002; 961:
372385.
171. Parikh SA and Edelman ER. Endothelial cell delivery
for cardiovascular therapy. Adv Drug Deliv Rev 2000;
42: 139161.
172. Jain RK. Molecular regulation of vessel maturation.
Nat Med 2003; 9: 685693.
173. Chen RR, Silva EA, Yuen WW and Mooney DJ.
Spatio-temporal VEGF and PDGF delivery patterns
blood vessel formation and maturation. Pharm Res
2007; 24: 258264.
174. Darland DC and DAmore PA. Blood vessel maturation: vascular development comes of age. J Clin Invest
1999; 103: 157158.
175. De la Riva B, Nowak C, Sanchez E, Hernandez A,
Schulz-Siegmund M, Pec MK, et al. VEGF-controlled

Downloaded from jba.sagepub.com at University of Liverpool on March 27, 2016

Naderi et al.

176.

177.

178.

179.

180.

181.

182.

183.

184.

185.

186.

187.
188.

189.

17

release within a bone defect from alginate/chitosan/PLAH scaffolds. Eur J Pharm Biopharm 2009; 73: 5058.
Sagnella S, Anderson E, Sanabria N, Marchant RE and
Kottke-Marchant K. Human endothelial cell interaction
with biomimetic surfactant polymers containing peptide
ligands from the heparin binding domain of fibronectin.
Tissue Eng 2005; 11: 226236.
Saltzman WM and Olbricht WL. Building drug delivery
into tissue engineering. Nat. Rev Drug Discov 2002; 1:
177186.
Kaigler D, Wang Z, Horger K, Mooney DJ and
Krebsbach PH. VEGF scaffolds enhance angiogenesis
and bone regeneration in irradiated osseous defects. J
Bone Miner Res 2006; 21: 735744.
Ennett AB, Kaigler D and Mooney DJ. Temporally regulated delivery of VEGF in vitro and in vivo. J Biomed
Mater Res 2006; 79A: 176184.
Briganti E, Spiller D, Mirtelli C, Kull S, Counoupas C,
Losi P, et al. A composite fibrin-based scaffold for
controlled delivery of bioactive pro-angiogenetic
growth factors. J Control Release 2010; 142: 1421.
Mikos AG, Herring SW, Ochareon P, Elisseeff J, Lu
HH, Kandel R, et al. Engineering complex tissues.
Tissue Eng 2006; 12: 33073339.
Shen YH, Shoichet MS and Radisic M. Vascular endothelial growth factor immobilized in collagen scaffold
promotes penetration and proliferation of endothelial
Cells. Acta Biomater 2008; 4: 477489.
Yang F, Cho SW, Son SM, Bogatyrev SR, Singh D,
Green JJ, et al. Genetic engineering of human stem
cells for enhanced angiogenesis using biodegradable
polymeric nanoparticles. Proc Nat Acad Sci USA
2010; 107: 33173322.
Vallbacka JJ and Sefton MV. Vascularization and
improved in vivo survival of VEGF-secreting cells microencapsulated in HEMA-MMA. Tissue Eng 2007; 13:
22592269.
Matsumoto T, Kuroda R, Mifune Y, Kawamoto A,
Shoji T, Miwa M, et al. Circulating endothelial/skeletal
progenitor cells for bone regeneration and healing. Bone
2008; 43: 434439.
Kulkarni SS, Orth R, Ferrari M and Moldovan NI.
Micropatterning of endothelial cells by guided stimulation with angiogenic factors. Biosens Bioelectron 2004;
19: 14011407.
Folkman J and DAmore PA. Blood vessel formation:
what is its molecular basis? Cell 1996; 87: 11531155.
Fahmy TM, Samstein RM, Harness CC and Mark
Saltzman W. Surface modification of biodegradable
polyesters with fatty acid conjugates for improved
drug targeting. Biomaterials 2005; 26: 57275736.
Davis ME, Hsieh PC, Takahashi T, Song Q, Zhang
S, Kamm RD, et al. Local myocardial insulin-like

190.

191.

192.

193.

194.

195.

196.

197.

198.

199.
200.

201.
202.

203.

204.

growth factor 1 (IGF-1) delivery with biotinylated


peptide nanofibers improves cell therapy for myocardial infarction. Proc Nat Acad Sci USA 2006; 103:
81558160.
Clapper JD, Pearce ME, Guymon CA and Salem AK.
Biotinylated biodegradable nanotemplated hydrogel
networks
for
cell
interactive
applications.
Biomacromolecules 2008; 9: 11881194.
Nomi M, Miyake H, Sugita Y, Fujisawa M and Soker S.
Role of growth factors and endothelial cells in therapeutic angiogenesis and tissue engineering. Curr Stem Cell
Res Ther 2006; 1: 333343.
Ennett AB, Kaigler D and Mooney DJ. Temporally regulated delivery of VEGF in vitro and in vivo. J Biomed
Mater Res 2006; 79A: 176184.
Ennett AB and Mooney DJ. Tissue engineering strategies for in vivo neovascularisation. Expert Opin Biol Ther
2002; 2: 805818.
Edelman ER, Mathiowitz E, Langer R and Klagsbrun
M. Controlled and modulated release of basic fibroblast
growth factor. Biomaterials 1991; 12: 619626.
Lavik E and Langer R. Tissue engineering: current
state and perspectives. Appl Microbiol Biotechnol 2004;
65: 18.
Storrie H and Mooney DJ. Sustained delivery of plasmid DNA from polymeric scaffolds for tissue engineering. Adv Drug Deliv Rev 2006; 58: 500514.
Segura T, Chung PH and Shea LD. DNA delivery
from hyaluronic acid-collagen hydrogels via a substrate-mediated approach. Biomaterials 2005; 26:
15751584.
Sheridan MH, Shea LD, Peters MC and Mooney DJ.
Bioabsorbable polymer scaffolds for tissue engineering
capable of sustained growth factor delivery. J Control
Release 2000; 64: 91102.
Stoop R. Smart biomaterials for tissue engineering of
cartilage. Injury 2008; 39: S77S87.
Rosso F, Marino G, Giordano A, Barbarisi M,
Parmeggiani D and Barbarisi A. Smart materials as
scaffolds for tissue engineering. J Cell Physiol 2005;
203: 465470.
Williams D. Environmentally smart polymers. Med
Device Technol 2005; 16: 913.
Lutolf MR, Weber FE, Schmoekel HG, Schense JC,
Kohler T, Muller R, et al. Repair of bone defects
using synthetic mimetics of collagenous extracellular
matrices. Nat Biotechnol 2003; 21: 513518.
Patel ZS and Mikos AG. Angiogenesis with biomaterialbased drug- and cell-delivery systems. J Biomater Sci
Polym Ed 2004; 15: 701726.
Fournier N and Doillon CJ. Biological moleculeimpregnated polyester: an in vivo angiogenesis study.
Biomaterials 1996; 17: 16591665.

Downloaded from jba.sagepub.com at University of Liverpool on March 27, 2016

You might also like